Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Mol Biol Rep ; 48(9): 6313-6321, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34390443

ABSTRACT

BACKGROUNDS: Epithelial mesenchymal transition (EMT) is a critical process involved in the invasion and metastasis of cancer, including lung cancer (LC). Transforming growth factor (TGF)-ß is one of factors capable of inducing EMT. Polyinosinic-polycytidylic acid (polyI:C), a synthetic agonist for toll-like receptor (TLR) 3, can enhance immune responses and has been used as an adjuvant for cancer vaccines; however, it remains unclear whether it influences other process, such as EMT. In the present study, we examined the effects of polyI:C on TGF-ß-treated A549 human LC cells. METHODS AND RESULTS: By in vitro cell proliferation assay, polyI:C showed no effect on the growth of A549 cells treated with TGF-ß1 at the concentration range up to 10 µg/ml; however, it markedly suppressed the motility in a cell scratch and a cell invasion assay. By Western blotting, polyI:C dramatically decreased TGF-ß1-induced Ak strain transforming (Akt) phosphorylation and increased phosphatase and tensin homologue (PTEN) expression without affecting the Son of mothers against decapentaplegic (Smad) 3 phosphorylation or the expression level of E-cadherin, N-cadherin or Snail, indicating that polyI:C suppressed cell motility independently of the 'cadherin switching'. The Akt inhibitor perifosine inhibited TGF-ß1-induced cell invasion, and the PTEN-specific inhibitor VO-OHpic appeared to reverse the inhibitory effect of polyI:C. CONCLUSION: PolyI:C has a novel function to suppress the motility of LC cells undergoing EMT by targeting the phosphatidylinositol 3-kinase/Akt pathway partly via PTEN and may prevent or reduce the metastasis of LC cells.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Cell Movement/drug effects , Lung Neoplasms/metabolism , Poly I-C/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung Neoplasms/pathology , Organometallic Compounds/pharmacology , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Recombinant Proteins/pharmacology , Toll-Like Receptor 3/agonists
2.
Biol Pharm Bull ; 43(10): 1511-1518, 2020.
Article in English | MEDLINE | ID: mdl-32999161

ABSTRACT

Gallbladder carcinoma (GBC) is one of the most common carcinomas of the biliary tract and is associated with aggressive malignancy and poor prognosis. Current therapeutic strategies, including surgery, radiotherapy, and chemotherapy, are not sufficient for the treatment of GBC, and new therapeutic strategies are urgently needed. The antitumor effects of oroxylin A (OrA), a natural flavonoid extracted from the dried roots of medicinal plants such as Scutellariae species (Radix Scutellariae), have been widely reported in various cancers. In this study, we first evaluated the antitumor activity and the underlying mechanism of action of OrA on GBC cells in vitro. Our results revealed that OrA significantly attenuated the proliferation, migration, and invasion of GBC cells, simultaneously promoting their apoptosis. Suppression of the phosphate on and tension homology deleted chromosome ten (PTEN)/phosphatidylinositol-3 kinase (PI3K)/protein kinase B (AKT) signaling pathway was found to be the underlying mechanism involved in the antitumor activity of OrA. In addition, experiments using a tumor xenograft mouse model confirmed the antitumor effects of OrA in vivo. Taken together, our findings indicate that OrA could be a potential antitumor agent for the prospective treatment of GBC.


Subject(s)
Antineoplastic Agents/therapeutic use , Flavonoids/therapeutic use , Gallbladder Neoplasms/drug therapy , PTEN Phosphohydrolase/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Flavonoids/pharmacology , Gallbladder Neoplasms/metabolism , Humans , Male , Mice , Mice, Nude , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Xenograft Model Antitumor Assays/methods
3.
Colloids Surf B Biointerfaces ; 184: 110499, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31541893

ABSTRACT

The invasive spreading of residual osteosarcoma cells becomes a serious threat to human health, urgently needing new bone regenerative biomaterials for orthopedic therapy. Thus, in this work, selenite-substituted hydroxyapatite (SeHA) nanoparticles were prepared for both inhibiting the recurrence of the tumor and accelerating the regenerative repair of bone defect. Physicochemical characterization showed these synthetic nanoparticles were spherical poly-crystals with the shape of snowflakes. Such structure benefited them to inhibit the cellular viability of osteosarcoma cells by about (58.90 ± 14.37)% during 24 h co-culturing. The expression level of cell growth-related genes such as PTEN, MMP-9, Cyclin D1, Cyclin A2, Annexin A2 and CDC2 decreased. Bisulfite Sequence PCR of PTEN gene exhibited about (22.40 ± 5.39)%, (45.91 ± 6.36)% and (25.90 ± 5.36)% promoter methylation in control, HA and SeHA group. Animal experiment also proved the similar effects. Almost no recurrence were observed in SeHA group. Oppositely, the slowly recurrent growth of the remnant tumor appeared in purely surgical group. The overall survival and toxicity analysis showed that, in the usage dose of 0-0.1 g, the SeHA-0.01 exhibited higher inhibitory recurrence and metastasis potentials, lower renal toxicity and better anti-inflammation function. Immunohistochemistry stain showed the reduced expression of PTEN, MMP-9, Ki-67 and Annexin A2, but slightly increased expression of DNMT1 and BMP-2. Compared the methylation status of PTEN gene in each group, it was confirming that SeHA nanoparticles hardly possessed the de-methylation effect, but the pure HA strikingly increased the methylation level of such gene. It seemed the dopant selenite ions possessed de-methylation effect onto PTEN gene. Therefore, from the viewpoint of inhibiting metastatic potentials, the SeHA-0.01 might be a feasible biomaterial to inhibit the relapse of the tumor post-surgery.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Durapatite/pharmacology , Nanoparticles/chemistry , Osteosarcoma/drug therapy , PTEN Phosphohydrolase/antagonists & inhibitors , Selenium/pharmacology , Animals , Apoptosis/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Durapatite/chemistry , Humans , Male , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Particle Size , Selenium/chemistry , Surface Properties , Tumor Cells, Cultured
4.
J Alzheimers Dis ; 63(2): 445-455, 2018.
Article in English | MEDLINE | ID: mdl-29614669

ABSTRACT

Vascular dementia (VaD) is the second most common dementia worldwide. Unlike Alzheimer's disease, VaD does not yet have effective therapeutic drugs. Harpagoside is the most important component extracted from Harpagophytum procumbens, a traditional Chinese medicine that has been widely used. The neuroprotective effects of harpagoside have been studied in Aß- and MPTP-induced neurotoxicity. However, whether harpagoside is protective against VaD is not clear. In this study, with the use of chronic cerebral hypoperfusion rats, a well-known VaD model, we demonstrated that chronic administration (two months) of harpagoside was able to restore both the spatial learning/memory and fear memory impairments. Importantly, the protective effects of harpagoside were not due to alterations in the physiological conditions, metabolic parameters, or locomotor abilities of the rats. Meanwhile, we found that harpagoside suppressed the overactivation of PTEN induced by CCH by enhancing PTEN phosphorylation. Furthermore, harpagoside elevated the activity of Akt and inhibited the activity of GSK-3ß, downstream effectors of PTEN. Overall, our study suggested that harpagoside treatment might be a potential therapeutic drug targeting the cognitive impairments of VaD.


Subject(s)
Dementia, Vascular/drug therapy , Glycosides/pharmacology , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , PTEN Phosphohydrolase/antagonists & inhibitors , Pyrans/pharmacology , Animals , Dementia, Vascular/pathology , Dementia, Vascular/physiopathology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Learning/drug effects , Male , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , PTEN Phosphohydrolase/metabolism , Random Allocation , Rats, Wistar
5.
Mol Cell ; 69(6): 923-937.e8, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29547721

ABSTRACT

Dietary supplements such as vitamins and minerals are widely used in the hope of improving health but may have unidentified risks and side effects. In particular, a pathogenic link between dietary supplements and specific oncogenes remains unknown. Here we report that chondroitin-4-sulfate (CHSA), a natural glycosaminoglycan approved as a dietary supplement used for osteoarthritis, selectively promotes the tumor growth potential of BRAF V600E-expressing human melanoma cells in patient- and cell line-derived xenograft mice and confers resistance to BRAF inhibitors. Mechanistically, chondroitin sulfate glucuronyltransferase (CSGlcA-T) signals through its product CHSA to enhance casein kinase 2 (CK2)-PTEN binding and consequent phosphorylation and inhibition of PTEN, which requires CHSA chains and is essential to sustain AKT activation in BRAF V600E-expressing melanoma cells. However, this CHSA-dependent PTEN inhibition is dispensable in cancer cells expressing mutant NRAS or PI3KCA, which directly activate the PI3K-AKT pathway. These results suggest that dietary supplements may exhibit oncogene-dependent pro-tumor effects.


Subject(s)
Carcinogens/toxicity , Cell Transformation, Neoplastic/genetics , Chondroitin Sulfates/toxicity , Dietary Supplements/toxicity , Melanoma/chemically induced , Mutation , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/chemically induced , Animals , Antinematodal Agents/pharmacology , Casein Kinase II/metabolism , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , GTP Phosphohydrolases/genetics , HEK293 Cells , HT29 Cells , Humans , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, Nude , Mice, Transgenic , NIH 3T3 Cells , Nuclear Proteins/genetics , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Transcription Factors/genetics , Xenograft Model Antitumor Assays
6.
Oncol Rep ; 37(2): 1011-1019, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28075472

ABSTRACT

The aim of the present study was to evaluate the effects of PTEN/AKT signaling on TUBB3 and TOP2A expression and on the subsequent cell growth of human breast cancer MCF-7 cells. We found that the disease-free survival (DFS) and overall survival (OS) of breast cancer patients with TUBB3­positive tumors were lower than these rates in the patients with TUBB3-negative tumors. Meanwhile, DFS and OS of breast cancer patients with TOP2A-positive tumors were also lower than these rates in patients with TOP2A-negative tumors. Suppression of PTEN reduced the protein expression of TUBB3 and TOP2A in MCF-7 cells. Suppression of PTEN also reduced cell proliferation and induced apoptosis and caspase-3 activity in MCF-7 cells. Moreover, an increase in ATP also reduced TUBB3 and TOP2A protein expression, reduced cell proliferation and induced apoptosis and caspase-3 activity in the MCF-7 cells following suppression of PTEN. Suppression of phosphorylation-AKT (p-AKT) reduced the protein expression of TUBB3 and TOP2A in the MCF-7 cells. Suppression of p-AKT also reduced cell proliferation and induced apoptosis and caspase-3 activity in the MCF-7 cells. Then, ATP also reduced TUBB3 and TOP2A protein expression, reduced cell proliferation and induced apoptosis and caspase-3 activity in MCF-7 cells following suppression of p-AKT. These results suggest that PTEN/AKT signaling affects the expression of TUBB3 and TOP2A reducing cell growth and inducing apoptosis of human breast cancer MCF-7 cells through ATP and caspase-3 signaling pathways. TUBB3 and TOP2A may be promising prognostic markers for the efficacy of adjuvant cisplatin-based chemotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Breast Neoplasms/pathology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Tubulin/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Antigens, Neoplasm/genetics , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Caspase 3/metabolism , Cell Proliferation , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Disease-Free Survival , Female , Humans , MCF-7 Cells , Organometallic Compounds/pharmacology , PTEN Phosphohydrolase/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tubulin/genetics , bcl-2-Associated X Protein/metabolism
7.
Neoplasma ; 63(3): 351-61, 2016.
Article in English | MEDLINE | ID: mdl-26925795

ABSTRACT

Epithelial-mesenchymal transition (EMT) is considered as the most important mechanism that underlies the initiation of cancer metastasis. Here we report that Physicon 8-O-ß-glucopyranoside (PG), a major active ingredient from a traditional Chinese herbal medicine Rumex japonicus Houtt, is capable of preventing human colorectal cancer cells from hypoxia-induced EMT. The treatment of the cells with PG reversed the EMT-related phenotype that has the morphological changes, down-regulation of E-cadherin, and hypoxia-induced cell migration and invasion. The effect was mediated at least in part by inhibiting the mRNA and protein expressions of EMMPRIN via modulation of PTEN/Akt/HIF-1α pathway. In addition, we found that PG-mediated prevention of EMT involved blockade of the hypoxia-induced up-regulation of Snail, Slug and Twist. In summary, this study showed that PG can prevent EMT induced by hypoxia, the environment that commonly exists in the center of a solid tumor. Given the low toxicity of PG to the healthy tissues, our study suggests that PG can serve as a safe therapeutic agent for suppressing cancer metastasis.


Subject(s)
Basigin/antagonists & inhibitors , Cell Hypoxia/drug effects , Colorectal Neoplasms/drug therapy , Emodin/analogs & derivatives , Epithelial-Mesenchymal Transition/drug effects , Glucosides/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Basigin/biosynthesis , Basigin/genetics , Cell Movement/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Emodin/pharmacology , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction/drug effects
8.
Oncogene ; 35(34): 4518-28, 2016 08 25.
Article in English | MEDLINE | ID: mdl-26853465

ABSTRACT

Mutations to fibroblast growth factor receptor 3 (FGFR3) and phosphatase and tensin homologue (PTEN) signalling pathway components (for example, PTEN loss, PIK3CA, AKT1, TSC1/2) are common in bladder cancer, yet small-molecule inhibitors of these nodes (FGFR/PTENi) show only modest activity in preclinical models. As activation of autophagy is proposed to promote survival under FGFR/PTENi, we have investigated this relationship in a panel of 18 genetically diverse bladder cell lines. We found that autophagy inhibition does not sensitise bladder cell lines to FGFR/PTENi, but newly identify an autophagy-independent cell death synergy in FGFR3-mutant cell lines between mTOR (mammalian target of rapamycin) pathway inhibitors and chloroquine (CQ)-an anti-malarial drug used as a cancer therapy adjuvant in over 30 clinical trials. The mechanism of synergy is consistent with lysosomal cell death (LCD), including cathepsin-driven caspase activation, and correlates with suppression of cSREBP1 and cholesterol biosynthesis in sensitive cell lines. Remarkably, loss of viability can be rescued by saturating cellular membranes with cholesterol or recapitulated by statin-mediated inhibition, or small interfering RNA knockdown, of enzymes regulating cholesterol metabolism. Modulation of CQ-induced cell death by atorvastatin and cholesterol is reproduced across numerous cell lines, confirming a novel and fundamental role for cholesterol biosynthesis in regulating LCD. Thus, we have catalogued the molecular events underlying cell death induced by CQ in combination with an anticancer therapeutic. Moreover, by revealing a hitherto unknown aspect of lysosomal biology under stress, we propose that suppression of cholesterol metabolism in cancer cells should elicit synergy with CQ and define a novel approach to future cancer treatments.


Subject(s)
Chloroquine/pharmacology , Cholesterol/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , Atorvastatin/pharmacology , Autophagy , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Drug Synergism , Humans , Lysosomes/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , TOR Serine-Threonine Kinases/physiology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
9.
Cell Physiol Biochem ; 36(3): 956-65, 2015.
Article in English | MEDLINE | ID: mdl-26087719

ABSTRACT

BACKGROUND: Berberine, a well-known component of the Chinese herbal medicine Huanglian, has wide range of biochemical and pharmacological effects, including antineoplastic effect, but the exact mechanisms remain unclear. The aim of the present study was to evaluate the potential chemo-sensitization effect of berberine in ovarian cancer cell line A2780. METHODS: The expression of miR-93 was measure by RT-PCR. The target of miR-93 was confirmed by luciferase activity assay. Hoechst 33258 staining, Annexin V and PI double staining were used for apoptosis analysis. RESULTS: In this study, we found A2780/DDP cells that were incubated with berberine combined with cisplatin had a significantly lower survival than the control group. Berberine enhanced cisplatin induced apoptosis and induced G0/G1 cell cycle arrest in A2780 cells. Next, we observed that the miR-93 levels in cisplatin resistant cell lines were higher than that in cisplatin sensitive cell lines. Furthermore, our study found berberine could inhibit miR-93 expression and function in ovarian cancer, as shown by an increase of its target PTEN, an important tumor suppressor in ovarian cancer. A2780 cells that were treated with PTEN siRNA had increased survival compared to NC group and this could be partly alleviated by the AKT inhibitor Triciribine. More importantly, A2780 cells that were treated with PTEN siRNA had a survival pattern that is similar to cells with miR-93 overexpression. CONCLUSION: The results suggested that berberine modulated the sensitivity of cisplatin through miR-93/PTEN/AKT signaling pathway in the ovarian cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Berberine/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Synergism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , PTEN Phosphohydrolase/agonists , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribonucleosides/pharmacology , Signal Transduction
10.
Neuroscience ; 292: 81-9, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25724866

ABSTRACT

Ginseng serves as a potential candidate for the treatment of aging-related memory decline or memory loss. However, the related mechanism is not fully understood. In this study, we applied an intraperitoneal injection of ginsenoside Rg1, an active compound from ginseng in middle-aged mice and detected memory improvement and the underlying mechanisms. Our results showed that a period of 30-day administration of ginsenoside Rg1 enhanced long-term memory in the middle-aged animals. Consistent with the memory improvement, ginsenoside Rg1 administration facilitated weak theta-burst stimulation (TBS)-induced long-term potentiation (LTP) in acute hippocampal slices from middle-aged animals. Ginsenoside Rg1 administration increased the dendritic apical spine numbers and area in the CA1 region. In addition, ginsenoside Rg1 administration up-regulated the expression of hippocampal p-AKT, brain-derived neurotrophic factor (BDNF), proBDNF and glutamate receptor 1 (GluR1), but not p-ERK. Interestingly, the phosphatase and tensin homolog deleted on chromosome ten (PTEN) inhibitor (bpV) mimicked the ginsenoside Rg1 effects, including increasing p-AKT expression, promoting hippocampal basal synaptic transmission, LTP and memory. Taken together, our data suggest that ginsenoside Rg1 treatment improves memory in middle-aged mice possibly through regulating the PI3K/AKT pathway, altering apical spines and facilitating hippocampal LTP.


Subject(s)
Ginsenosides/pharmacology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Memory, Long-Term/drug effects , Nootropic Agents/pharmacology , Aging/drug effects , Aging/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dendritic Spines/drug effects , Dendritic Spines/physiology , Electric Stimulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Fear/drug effects , Fear/physiology , Hippocampus/physiology , Long-Term Potentiation/physiology , Male , Memory, Long-Term/physiology , Mice, Inbred C57BL , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, AMPA/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tissue Culture Techniques
11.
PLoS One ; 9(6): e100880, 2014.
Article in English | MEDLINE | ID: mdl-24978597

ABSTRACT

Tumor cells upregulate many cell signaling pathways, with AKT being one of the key kinases to be activated in a variety of malignancies. GSK2110183 and GSK2141795 are orally bioavailable, potent inhibitors of the AKT kinases that have progressed to human clinical studies. Both compounds are selective, ATP-competitive inhibitors of AKT 1, 2 and 3. Cells treated with either compound show decreased phosphorylation of several substrates downstream of AKT. Both compounds have desirable pharmaceutical properties and daily oral dosing results in a sustained inhibition of AKT activity as well as inhibition of tumor growth in several mouse tumor models of various histologic origins. Improved kinase selectivity was associated with reduced effects on glucose homeostasis as compared to previously reported ATP-competitive AKT kinase inhibitors. In a diverse cell line proliferation screen, AKT inhibitors showed increased potency in cell lines with an activated AKT pathway (via PI3K/PTEN mutation or loss) while cell lines with activating mutations in the MAPK pathway (KRAS/BRAF) were less sensitive to AKT inhibition. Further investigation in mouse models of KRAS driven pancreatic cancer confirmed that combining the AKT inhibitor, GSK2141795 with a MEK inhibitor (GSK2110212; trametinib) resulted in an enhanced anti-tumor effect accompanied with greater reduction in phospho-S6 levels. Taken together these results support clinical evaluation of the AKT inhibitors in cancer, especially in combination with MEK inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Diamines/pharmacology , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Tumor Burden/drug effects , Administration, Oral , Animals , Antineoplastic Agents/chemical synthesis , Blood Glucose/metabolism , Cell Line, Tumor , Diamines/chemical synthesis , Drug Evaluation, Preclinical , Drug Synergism , Female , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, SCID , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/chemical synthesis , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
12.
J Ethnopharmacol ; 155(1): 362-72, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24882728

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Sarcopoterium spinosum is an abundant plant in Israel, used by Bedouin medicinal practitioners for the treatment of diabetes. In our previous study we validated the anti-diabetic activity of Sarcopoterium spinosum. The aim of this study was to further clarify its mechanism of action. MATERIALS AND METHODS: In-vivo studies were performed on KK-a/y mice given the extract for 6 weeks. Insulin tolerance test was performed, and relative pancreatic islets area was measured. Mechanisms of action were investigated in L6 myotubes using protein array, Western blot analysis and confocal microscopy. Glucose uptake assays were performed in 3T3-L1 adipocytes. RESULTS: Sarcopoterium spinosum extract reduced fasting blood glucose and improved insulin sensitivity in treated mice. Hypertrophic islets were detected in diabetic, but not in Sarcopoterium spinosum-treated mice. Sarcopoterium spinosum phosphorylated PTEN on ser380 and thr382/383, which are known inhibitory sites. PKB was not phosphorylated by Sarcopoterium spinosum, however, translocation of PKB from cytoplasm to the membrane and nucleus was detected. Target proteins of PKB were regulated by Sarcopoterium spinosum; GSK3ß was phosphorylated and cytosolic localization of FoxO was increased. Glucose uptake was increased in a PI3K and AMPK-independent mechanism. CONCLUSIONS: We suggest that Sarcopoterium spinosum inhibited PTEN and activated PKB by a mechanism which is independent of ser473 and thr308 phosphorylation. Other post translation modifications might be involved and should be analyzed further in order to understand this unique PKB activation. Identifying the active molecules in the extract, may lead to the development of new agents for the treatment of insulin resistance.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Insulin/metabolism , Plant Extracts/pharmacology , Rosaceae/chemistry , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Glucose/metabolism , Insulin Resistance , Israel , Male , Medicine, Traditional , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism
13.
Mol Hum Reprod ; 20(8): 736-44, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24830779

ABSTRACT

In the mammalian ovary a small number of follicles are steadily recruited from the quiescent pool to undergo development. Follicle loss, maintenance and growth are strictly controlled by complex molecular interactions including the phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt) signalling pathway. Stimulation of PI3K promotes phosphorylation of Akt resulting in follicle survival and activation of growth whereas this pathway is suppressed by the actions of the phosphatase and tensin homologue (PTEN). The aim of this study was to determine the effect of dipotassium bisperoxo(5-hydroxypyridine-2-carboxyl)oxovanadate (bpV), a reversible inhibitor of PTEN, on the activation, survival and development of human ovarian follicles in vitro. Biopsied ovarian tissue fragments were obtained from 17 women aged 23-46 years and exposed to 1 µM bpV(HOpic) (n = 146) or control medium (n = 128) for 24 h. Media were then replaced with control medium and all tissue incubated for a further 5 days. Ovarian tissue from each treatment group was fixed after the initial 24 h culture period and phosphorylated Akt was quantified by western blotting. After 6 days incubation all tissue fragments were inspected under light microscopy and any secondary follicles ≥100 µm isolated. Isolated follicles were cultured individually in control medium supplemented with 100 ng/ml recombinant human activin A. Tissue fragments without follicles suitable for isolation were fixed and processed for histological and immunohistochemical analysis. During 6 days culture, follicle activation occurred in tissue samples from both treatment groups but with significantly more follicles progressing to the secondary stage of development in the presence of 1 µM bpV(HOpic) compared with control (31 versus 16%; P < 0.05). Increased activation was associated with increased Akt phosphorylation and increased nuclear export of FOXO3. However isolated and cultured follicles that had been exposed to bpV(HOpic) showed limited growth and reduced survival compared with follicles from control fragments (P < 0.05). This study demonstrates that inhibition of PTEN with bpV(HOpic) affects human ovarian follicle development by promoting the initiation of follicle growth and development to the secondary stage, as in rodent species, but severely compromises the survival of isolated secondary follicles.


Subject(s)
Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovary/growth & development , Ovary/metabolism , PTEN Phosphohydrolase/metabolism , Adult , Blotting, Western , Female , Humans , Immunohistochemistry , Middle Aged , Ovarian Follicle/drug effects , Ovary/drug effects , PTEN Phosphohydrolase/antagonists & inhibitors , Tissue Culture Techniques , Vanadium Compounds/pharmacology , Young Adult
14.
Metallomics ; 6(7): 1277-87, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24759986

ABSTRACT

Free zinc ions (Zn(2+)) participate in several signaling pathways. The aim of the present study was to investigate a potential involvement of Zn(2+) in the PI3K/Akt pathway of interleukin (IL)-2 signaling in T-cells. The IL-2 receptor triggers three major pathways, ERK1/2, JAK/STAT5, and PI3K/Akt. We have previously shown that an IL-2-mediated release of lysosomal Zn(2+) into the cytoplasm activates ERK1/2, but not STAT5. In the present study, Akt phosphorylation in response to IL-2 was abrogated by the Zn(2+) chelator N,N,N',N'-tetrakis-2(pyridyl-methyl)ethylenediamine, and was induced by treatment with Zn(2+) and the ionophore pyrithione. The latter were ineffective in cells that were treated with siRNA against the phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a phosphatase that degrades the lipid second messenger PI(3,4,5)P3, which is produced by PI3K and leads to activation of Akt. Inhibition of recombinant PTEN by Zn(2+)in vitro yielded an IC50 of 0.59 nM. Considering a resting free cytoplasmic Zn(2+) level of 0.2 nM in the T-cell line CTLL-2, this seems ideally suited for dynamic regulation by cellular Zn(2+). Oxidation with H2O2 and supplementation with Zn(2+) led to similar changes in the CD spectrum of PTEN. Moreover, Zn(2+) partially prevented the oxidation of cysteines 71 and 124. Hence, we hypothesize that zinc signals affect the IL-2-dependent PI3K/Akt pathway by inhibiting the negative regulator PTEN through binding with a sub-nanomolar affinity to cysteine residues that are essential for its catalytic activity.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-2/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Zinc/pharmacology , Animals , Humans , Interleukin-2/pharmacology , Jurkat Cells , Male , Phosphorylation , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
15.
Int J Mol Sci ; 14(10): 19792-804, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24084726

ABSTRACT

Achyranthes bidentata, a Chinese medicinal herb, is reported to be neuroprotective. However, its role in cardioprotection remains largely unknown. Our present study aimed to investigate the effects of Achyranthes bidentata polypeptides (ABPP) preconditioning on myocardial ischemia/reperfusion (MI/R) injury and to test the possible mechanisms. Rats were treated with ABPP (10 mg/kg/d, i.p.) or saline once daily for one week. Afterward, all the animals were subjected to 30 min of myocardial ischemia followed by 4 h of reperfusion. ABPP preconditioning for one week significantly improved cardiac function following MI/R. Meanwhile, ABPP reduced infarct size, plasma creatine kinase (CK)/lactate dehydrogenase (LDH) activities and myocardial apoptosis at the end of reperfusion in rat hearts. Moreover, ABPP preconditioning significantly inhibited superoxide generation, gp91phox expression, malonaldialdehyde formation and enhanced superoxide dismutase activity in I/R hearts. Furthermore, ABPP treatment inhibited PTEN expression and increased Akt phosphorylation in I/R rat heart. PI3K inhibitor wortmannin blocked Akt activation, and abolished ABPP-stimulated anti-oxidant effect and cardioprotection. Our study demonstrated for the first time that ABPP reduces oxidative stress and exerts cardioprotection against MI/R injury in rats. Inhibition of PTEN and activation of Akt may contribute to the anti-oxidant capacity and cardioprotection of ABPP.


Subject(s)
Achyranthes/metabolism , Cardiotonic Agents/pharmacology , Heart/drug effects , Myocardial Reperfusion Injury/drug therapy , Oxidative Stress/drug effects , Peptides/pharmacology , Androstadienes/pharmacology , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Creatine Kinase/blood , L-Lactate Dehydrogenase/blood , Membrane Glycoproteins/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , NADPH Oxidase 2 , NADPH Oxidases/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Superoxides/metabolism , Wortmannin
16.
Am J Physiol Heart Circ Physiol ; 302(5): H1116-22, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22198171

ABSTRACT

Several studies have focused on the beneficial effects of peripheral angiotensin-(1-7) [Ang-(1-7)] in the regulation of cardiovascular function, showing its counterregulatory effect against the actions of angiotensin II (ANG II). However, its actions in the central nervous system are not completely understood. In the present study, we investigated the intracellular mechanisms underlying the action of ANG-(1-7) using the patch-clamp technique in neurons cultured from the hypothalamus of neonatal spontaneously hypertensive (SHR) and Wistar-Kyoto (WKY) rats. Superfusion of neurons with ANG II (100 nM) significantly increased neuronal firing in both strains of rats, and this chronotropic effect of ANG II was significantly enhanced in prehypertensive SHR neurons compared with WKY rat neurons. The enhanced chronotropic effect of ANG II was attenuated by a phosphatidylinositol 3-kinase (PI3-kinase) inhibitor, LY 294002 (10 µM). Superfusion of neurons with ANG-(1-7) (100 nM) did not alter the neuronal firing rate in either SHR or WKY neurons; however, it significantly attenuated the chronotropic action of ANG II exclusively in prehypertensive SHR neurons. This counterregulatory effect of ANG-(1-7) on ANG II action in prehypertensive SHR neurons was attenuated by cotreatment with either A-779, a Mas receptor antagonist, or bisperoxovanadium, a phosphatase and tensin homologue deleted on chromosome ten (PTEN) inhibitor. In addition, incubation of WKY and prehypertensive SHR neurons with ANG-(1-7) significantly increased PTEN activity. The data demonstrate that ANG-(1-7) counterregulates the chronotropic action of ANG II via a PTEN-dependent signaling pathway in prehypertensive SHR neurons.


Subject(s)
Angiotensin II/pharmacology , Angiotensin I/pharmacology , Angiotensinogen/pharmacology , Heart Rate/drug effects , PTEN Phosphohydrolase/metabolism , Peptide Fragments/pharmacology , Angiotensin I/antagonists & inhibitors , Angiotensin II/analogs & derivatives , Animals , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Hypothalamus/drug effects , Male , Morpholines/pharmacology , Neurons/drug effects , PTEN Phosphohydrolase/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Vanadium Compounds/pharmacology
17.
J Am Chem Soc ; 128(51): 16464-5, 2006 Dec 27.
Article in English | MEDLINE | ID: mdl-17177371

ABSTRACT

The activation of phosphatidylinositol 3-kinase (PI 3-K) and subsequent production of PtdIns(3,4,5)P3 launches a signal transduction cascade that impinges on a plethora of downstream effects on cell physiology. Control of PI 3-K and PtdIns(3,4,5)P3 levels is an important therapeutic target in treatments for allergy, inflammation, cardiovascular, and malignant human diseases. We designed metabolically stabilized, that is, phosphatase resistant, analogues of PtdIns(3,4,5)P3 as probes for long-lived potential agonists or potential antagonists for cellular events mediated by PtdIns(3,4,5)P3. In particular, two types of analogues were prepared containing phosphomimetics that would be selectively resistant to the lipid 3-phosphatase PTEN. The total asymmetric synthesis of the 3-phosphorothioate-PtdIns(3,4,5)P3 and 3-methylenephosphonate-PtdIns(3,4,5)P3 analogues is described. These two analogues showed differential binding to PtdIns(3,4,5)P3 binding modules, and both were potential long-lived activators that mimicked insulin action in sodium transport in A6 cells.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , PTEN Phosphohydrolase/antagonists & inhibitors , Phosphatidylinositol Phosphates/chemical synthesis , Phosphatidylinositol Phosphates/pharmacology , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Molecular Structure , Organophosphonates/chemistry , Phosphates/chemistry , Phosphatidylinositol Phosphates/chemistry , Structure-Activity Relationship , Time Factors
18.
EMBO J ; 25(11): 2377-87, 2006 Jun 07.
Article in English | MEDLINE | ID: mdl-16675953

ABSTRACT

In obesity and diabetes, the ability of hypothalamic neurons to sense and transduce changes in leptin and insulin levels is compromised. The effects of both hormones require intracellular signalling via the PI3-kinase pathway, which is inhibited by the phosphatase PTEN. We show that leptin-stimulated F-actin depolymerization in mouse hypothalamic cells is inhibited by PTEN, a process involving independent effects of both its lipid and protein phosphatase activities. Potentially mediating this F-actin depolymerization, leptin, but not insulin, stimulated the phosphorylation of PTEN in a CK2 dependent manner, and inhibited its phosphatase activity. Similarly, hyperpolarization of mouse pancreatic beta-cells by leptin also requires coincident PtdIns(3,4,5)P3 generation and actin depolymerization, and could be inhibited by mechanisms requiring both the lipid and protein phosphatase activities of PTEN. These results demonstrate a critical role for PTEN in leptin signalling and indicate a mechanism by which leptin and insulin can produce PI3K dependent differential cellular outputs.


Subject(s)
Hypothalamus/cytology , Insulin-Secreting Cells/metabolism , Leptin/metabolism , PTEN Phosphohydrolase/metabolism , Signal Transduction/physiology , Actins/metabolism , Animals , Cells, Cultured , Hypothalamus/metabolism , Insulin-Secreting Cells/cytology , Mice , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Patch-Clamp Techniques , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Potassium Channels/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Leptin
SELECTION OF CITATIONS
SEARCH DETAIL